Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Publication year range
1.
Cell Death Dis ; 4: e469, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23348583

ABSTRACT

The CD20-homolog Ms4a8a has recently been shown to be a marker for alternatively activated macrophages but its expression is not restricted to hematopoietic cells. Here, MS4A8A/MS4A8B expression was detected in differentiated intestinal epithelium in mouse and human, respectively. Interestingly, no MS4A8B expression was found in human colon carcinoma. Forced overexpression of MS4A8A in the murine colon carcinoma cell line CT26 led to a reduced proliferation and migration rate. In addition, MS4A8A-expressing CT26 cells displayed an increased resistance to hydrogen peroxide-induced apoptosis, which translated in an increased end weight of subcutaneous MS4A8A+ CT26 tumors. Gene profiling of MS4A8A+ CT26 cells revealed a significant regulation of 225 genes, most of them involved in cytoskeletal organization, apoptosis, proliferation, transcriptional regulation and metabolic processes. Thereby, the highest upregulated gene was the intestinal differentiation marker cytokeratin 20. In conclusion, we show that MS4A8A/MS4A8B is a novel differentiation marker of the intestinal epithelium that supports the maintenance of a physiological barrier function in the gut by modulating the transcriptome and by conferring an increased resistance to reactive oxygen species. The absence of MS4A8B in human colonic adenocarcinomas shown in this study might be a helpful tool to differentiate between healthy and neoplastic tissue.


Subject(s)
Epithelial Cells/metabolism , Membrane Proteins/metabolism , Animals , Apoptosis/drug effects , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytoskeleton , Gene Expression Profiling , Humans , Hydrogen Peroxide/toxicity , Intestinal Mucosa/pathology , Keratin-20/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism
2.
J Pathol ; 209(1): 67-77, 2006 May.
Article in English | MEDLINE | ID: mdl-16482496

ABSTRACT

Lymphangiogenesis is a novel prognostic parameter for several cancers that is preferentially quantified by immunohistochemistry of the lymphatic endothelium-specific hyaluronan receptor LYVE-1. Recently, the specificity of LYVE-1 was challenged by serendipitous observations of LYVE-1 expression in rare tissue macrophages. As expression of the hyaluronan receptor-like molecule stabilin-1 is shared by sinusoidal endothelium and macrophages, a thorough analysis of LYVE-1 expression was performed using macrophage-specific markers in vivo and in vitro. In murine tumour models and excisional wound healing, LYVE-1 expression occurred in a subset of CD11b(+), F4/80(+) tissue macrophages that preferentially co-expressed stabilin-1. Upon comparison of single- and double-labelling immunofluorescence, it became apparent that LYVE-1(+) macrophages mimic sprouting and collapsed lymphatic vessels. In vitro, LYVE-1 expression was induced in 25-40% of murine bone marrow-derived macrophages upon exposure to B16F1 melanoma-conditioned medium and IL-4/dexamethasone. By FACS analysis, 11.5% of bone marrow-derived macrophages were LYVE-1(+), stabilin-1(+) double-positive, while 9.9% were LYVE-1(+), stabilin-1(-) and 33.5% were LYVE-1(-), stabilin-1(+). Northern and western analyses confirmed expression of LYVE-1 mRNA and protein in bone marrow-derived macrophages. In the light of the current debate about true endothelial trans-differentiation versus endothelial mimicry of monocytes/macrophages, LYVE-1(+), stabilin-1(+) non-continuous endothelial-like macrophages will require further developmental and functional analyses. In conclusion, the findings imply that LYVE-1 staining must be supplemented by double labelling with macrophage markers in order to differentiate clearly between LYVE-1(+) lymphatics and LYVE-1(+) tumour-infiltrating macrophages. This improved approach will help to clarify the prognostic significance of lymphangiogenesis in malignant tumours.


Subject(s)
Endothelium, Lymphatic/metabolism , Glycoproteins/metabolism , Lymphangiogenesis/physiology , Macrophages/metabolism , Melanoma/metabolism , Animals , Antigens, Differentiation/analysis , Bone Marrow Cells/metabolism , CD11b Antigen/analysis , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Disease Models, Animal , Female , Macrophages/physiology , Melanoma/pathology , Melanoma/secondary , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Neoplasm Proteins/metabolism , Tumor Cells, Cultured , Vesicular Transport Proteins , Wound Healing/physiology
3.
J Pathol ; 208(4): 574-89, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16440291

ABSTRACT

Sentinel lymph node biopsy for several cancers has shown that metastatic tumour cells are preferentially arrested in the lymph node sinuses. To study the molecular components of this sinusoidal trap, gene profiling of lymph node (sinuses) versus tonsil (no sinuses) was performed. Among other groups of molecules, an intriguing gene signature of scavenger and lectin-like receptors was identified. Nine of the 13 genes were preferentially expressed in sinusoidal cells by immunohistochemistry. Using stabilin-2 and monoclonal antibody 3A5 as exclusive endothelial cell (EC) and macrophage (Mvarphi) markers, respectively, lymph node sinusoidal ECs (stabilin-2+, LYVE-1+, DC-SIGNR+, MARCO+, stabilin-1+, MMR+) and sinusoidal Mvarphi (MMR+, DC-SIGN+, sialoadhesin+, CD163+, stabilin-1+ ) showed distinct, but overlapping expression patterns of the signature molecules by double labelling immunofluorescence. The number of stabilin-1+ sinusoidal Mvarphi, however, varied considerably between samples, indicating turnover/differentiation dynamics in this sinusoidal cell population. In the hepatic sinuses, LYVE-1 and CD36 were strongly up-regulated on both sinusoidal ECs and Mvarphi, while DC-SIGNR and DC-SIGN were strongly down-regulated; in contrast to lymph node sinusoidal ECs, MARCO was confined to Mvarphi (Kupffer cells) in the liver sinuses. As Mvarphi are not present in the wall and lumen of splenic sinuses, splenic sinuses expressed a considerably reduced repertoire of scavenger/lectin receptors lacking sialoadhesin, CD36, CD163, and MARCO; in addition, DC-SIGNR was absent from splenic sinusoidal ECs, while DC-SIGN and thrombomodulin were strongly expressed. Interestingly, most of the signature molecules are known to mediate tumour cell adhesion in addition to their functions as scavenger or pattern recognition receptors. This study establishes a gene and tissue database platform to test the hypothesis that additive expression of the lymph node sinus signature genes in sinusoidal ECs and Mvarphi may contribute to selective tumour cell metastasis in lymph nodes and liver including organ-specific mechanisms, such as intraluminal retention or transmigration, while sparing the spleen.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Profiling , Lymph Nodes/metabolism , Lymphatic Metastasis , Macrophages/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Scavenger/genetics , Biomarkers/analysis , Cell Adhesion Molecules/genetics , Humans , Immunohistochemistry , Lectins/genetics , Liver/metabolism , Lymph Nodes/pathology , Microscopy, Confocal , Palatine Tonsil/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism
4.
Article in English | MEDLINE | ID: mdl-11586068

ABSTRACT

The Th1/Th2 paradigm has stimulated extensive research into the mechanisms underlying T-cell polarization; alternative activation of antigen-presenting cells (APCs) has turned out to be the corresponding concept APC polarization. Macrophages (M phi) as well as dendritic cells (DCs) can undergo Th1- or Th2-like polarization; APC1 and APC2 thus acquire the capacity to drive the development of naive T cells and the reactivation of resting T cells towards either a Th1 or a Th2 phenotype, respectively. Among polarized APC, effector macrophages are classically activated by mediators such as IFN-gamma, TNF-alpha or LPS (M phi 1), while M phi 2 are alternatively activated by IL-4, IL-10 or PGE(2). M phi 2 exhibit a unique molecular repertoire including receptors of innate immunity with broad specificity for foreign antigen and anti-inflammatory cytokines such as IL-1 receptor antagonist and alternative macrophage activation-associated CC-chemokine (AMAC)-1. While DC1 are well characterized, contradictory results have been obtained for DC2 that may either represent immature myeloid DCs or lymphoid DCs. Altogether, APC2 have come to age; they mediate Th2 differentiation, tolerance induction, downregulation of inflammation and healing. Thus, APC2 represent a hitherto neglected, but indispensable major pathway of APC activation and function.


Subject(s)
Antigen-Presenting Cells/immunology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/pathology , Humans , Skin/cytology , Skin/pathology , Skin Diseases/pathology
5.
Hautarzt ; 52(3): 193-200, 2001 Mar.
Article in German | MEDLINE | ID: mdl-11284063

ABSTRACT

Lymphocytes do not just act as immunological effector cells, but also play an important role in the regulation of the immune response. They are able to induce or suppress inflammatory reactions and this balancing function is reflected in the well-known Th1/Th2 concept. Lymphocytes depend on antigen presenting cells (APC) for induction of differentiation and specific activation mediated by antigen capture, processing and presentation. Thus, APC represent a link between innate and acquired immunity. In parallel to the Th1/Th2 dichotomy, APC may be subdivided into (a) pro-inflammatory, classically activated APC such as mature dendritic cells and IFN-gamma-activated effector macrophages, and (b) into anti-inflammatory, alternatively activated APC such as IL-10-activated immature dendritic cells and IL-4-induced suppressor macrophages. Alternatively activated APC may mediate induction and maintenance of tolerance towards allergens and environmental substances, control the course of inflammatory reactions, and participate in healing processes by enhancing angiogenesis. Malignant tumors and certain infectious agents may misuse alternatively activated APC for their purposes, thereby requiring counter-action by Th1 lymphocytes and classically activated APC. The concept of alternative activation thus confirms the important role of APC in maintaining the balance between induction and suppression of both inflammation and immunity and it opens new perspectives for the development of specific immunotherapeutic approaches.


Subject(s)
Antigen-Presenting Cells , Immune Tolerance , Macrophage Activation , Macrophages/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , Cytokines/immunology , Cytokines/physiology , Dermatitis, Contact/immunology , HIV Infections/immunology , Humans , Inflammation/immunology , Lymphocyte Activation , Macrophages/immunology , Melanoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Neoplasms/immunology , T-Lymphocytes/immunology
6.
Scand J Immunol ; 53(4): 386-92, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285119

ABSTRACT

Alternative activation of macrophages, induced by Th2 cytokines and glucocorticoids, is essential for the proper functioning of anti-inflammatory immune reactions. To this end, alternatively activated macrophages (aaMPhi) express a not yet fully unravelled set of genes including cytokines such as alternative macrophage activation-associated CC-chemokine (AMAC)-1 and pattern recognition molecules such as the scavenger receptor CD163. In order to further characterize the molecular repertoire of aaMPhi, differential gene expression was analyzed by combining subtractive suppression cloning and differential hybridization. We show here that aaMPhi induced by interleukin (IL)-4 overexpress the prototype extracellular matrix (ECM) protein fibronectin on the mRNA and protein level. This overall increase is accompanied by a shift in fibronectin splice variants from an embryonic to a mature pattern. In addition, the expression of another ECM protein, betaIG-H3, is also upregulated by IL-4 in aaMPhi. In contrast to IL-4 and in line with its inhibitory effect on wound healing, dexamethasone exerts a strongly suppressive effect on fibronectin and betaIG-H3 expression. In conclusion, overexpression of ECM proteins induced by IL-4 in macrophages suggests that aaMPhi may be involved in ECM deposition and tissue remodelling during the healing phase of acute inflammatory reactions and in chronic inflammatory diseases.


Subject(s)
Extracellular Matrix Proteins/genetics , Fibronectins/genetics , Macrophages/immunology , Macrophages/metabolism , Neoplasm Proteins/genetics , Transforming Growth Factor beta , Alternative Splicing , Base Sequence , DNA Primers/genetics , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Gene Expression , Humans , Immunohistochemistry , In Vitro Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Macrophage Activation , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
J Mol Evol ; 48(6): 770-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10229581

ABSTRACT

Eukaryotic vesicular transport requires the recognition of membranes through specific protein complexes. The heterotetrameric adaptor protein complexes 1, 2, and 3 (AP1/2/3) are composed of two large, one small, and one medium adaptin subunit. We isolated and characterized the cDNA for Arabidopsis gamma-adaptin and performed a phylogenetic analysis of all adaptin subunits (proteins) in the context of all known homologous proteins. This analysis revealed (i) that the large subunits of AP1/2/3 are homologous and (ii) two subunits of the heptameric coatomer I (COPI) complex belong to this gene family. In addition, all small subunits and the aminoterminal domain of the medium subunits of the heterotetramers are homologous to each other; this also holds for two corresponding subunits of the COPI complex. AP1/2/3 and a substructure (heterotetrameric, F-COPI subcomplex) of the heptameric COPI had a common ancestral complex (called pre-F-COPI). Since all large and all small/medium subunits share sequence similarity, the ancestor of this complex is inferred to have been a heterodimer composed of one large and one small subunit. The situation encountered today is the result of successive rounds of coordinated gene duplications of both the large and the small/medium subunits, with F-COPI being the first that separated from the ancestral pre-F-COPI.


Subject(s)
Coated Vesicles/metabolism , Eukaryotic Cells/metabolism , Evolution, Molecular , Membrane Proteins/genetics , Phylogeny , Adaptor Protein Complex 1 , Adaptor Protein Complex alpha Subunits , Adaptor Protein Complex gamma Subunits , Adaptor Proteins, Vesicular Transport , Arabidopsis/genetics , Arabidopsis/metabolism , Biological Transport , Coatomer Protein
8.
Pathobiology ; 67(5-6): 222-6, 1999.
Article in English | MEDLINE | ID: mdl-10725788

ABSTRACT

In parallel to the Th1/Th2 paradigm, antigen-presenting cells (APC) are divided into classically activated APC (dendritic cells/effector macrophages) and alternatively activated APC (IL-4-induced, alternatively activated macrophages/IL-10-induced, immature dendritic cells). Alternatively activated APC share a special molecular repertoire including receptors of innate immunity with broad specificity for foreign antigen and anti-inflammatory cytokines such as IL-1Ra and alternative macrophage activation-associated CC-chemokine-1. Alternatively activated APC mediated tolerance and downregulated inflammation. Abuse of alternatively activated APC in support of infectious susceptibility or tumor immune escape is counteracted by the classical pathway. Thus, classically and alternatively activated APC secure the balance between proinflammatory and anti-inflammatory immune reactions.


Subject(s)
Macrophage Activation/physiology , Macrophages/immunology , Animals , Antigen-Presenting Cells/immunology , Chemokines, CC/genetics , Chemokines, CC/metabolism , Dendritic Cells/immunology , Humans , Immune Tolerance/immunology , Macrophages/metabolism , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL